TIBS-1055; No. of Pages 9

Review

Emerging roles of the p38 MAPK and PI3K/AKT/mTOR pathways in oncogene-induced senescence Yingxi Xu1,2, Na Li1, Rong Xiang1, and Peiqing Sun2 1

College of Medicine, Nankai University, 94 Weijin Road, Tianjin, China, 300071 Department of Cell and Molecular Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA

2

Oncogene-induced senescence (OIS) is a tumor-suppressing response that must be disrupted for cancer to develop. Mechanistic insights into OIS have begun to emerge. Activation of the p53/p21WAF1 and/or p16INK4A tumor-suppressor pathways is essential for OIS. Moreover, the DNA damage response, chromatin remodeling, and senescence-associated secretory phenotype (SASP) are important for the initiation and maintenance of OIS. This review discusses recent advances in elucidating the mechanisms of OIS, focusing on the roles of the p38 mitogen-activated protein kinase (MAPK) and phosphoinositide 3-kinase (PI3K)/cellular homolog of murine thymoma virus AKT/mammalian target of rapamycin (mTOR) pathways. These studies indicate that OIS is mediated by an intricate signaling network. Further delineation of this network may lead to development of new cancer therapies targeting OIS. Introduction: OIS Replicative senescence is a form of irreversible growth arrest associated with the exhaustion of replicative potential of in vitro cultured cells [1]. In the case of human cells, replicative senescence occurs as a result of telomere erosion during cell division [2]. Replication-independent senescence can also be induced prematurely in young cells by activation of oncogenes. It was discovered in 1997 that in early-passaged normal human and murine fibroblasts, oncogenic ras induces an initial phase of hyperproliferation, followed by an irreversible growth arrest that is phenotypically indistinguishable from replicative senescence [3]. This form of premature senescence is termed oncogene-induced senescence, or OIS. The induction of OIS was initially reported to be independent of telomere length and telomerase activity [4], but a recent study has indicated that oncogenes such as ras induce telomere dysfunction, including telomere attrition in primary fibroblasts, and that OIS is not stable in cells with high telomerase activity [5]. Like replicative senescence, OIS is Corresponding author: Sun, P. ([email protected]). Keywords: oncogene-induced senescence; p38; phosphoinositide 3-kinase; AKT. 0968-0004/ ß 2014 Elsevier Ltd. All rights reserved. http://dx.doi.org/10.1016/j.tibs.2014.04.004

identified by senescence biomarkers such as senescenceassociated b-galactosidase (SA-b-gal). In addition to oncogenic ras, OIS can also be induced by other oncogenes, such as BRAF V600E, AKT, E2F1, cyclin E, mos, and Cdc6, and by inactivation of tumor-suppressor genes including PTEN and NF1 [6]. Like apoptosis, OIS is a tumor-suppressing defense mechanism that must be compromised by additional mutations during tumorigenesis. It has long been recognized that OIS inhibits oncogenic transformation in cell culture [7]. Later studies have demonstrated that OIS indeed occurs in multiple human tumor types and mouse cancer models, and serves as an initial barrier to cancer development in vivo [6]. The molecular mechanisms and signaling pathways that mediate OIS have begun to emerge [6]. Almost all the OIS inducers trigger activation of p53, which induces the expression of its transcriptional target p21WAF1, and/or increase the expression of p16INK4A [3]. p21WAF1 and/or p16INK4A both inhibit the activity of cyclin-dependent protein kinases (CDKs) that phosphorylate and inactivate the retinoblastoma protein (Rb), leading to accumulation of the hypophosphorylated, active form of Rb that mediates cellcycle arrest and other phenotypes of senescence. Some oncogenes induce OIS through DNA damage responses, which can be generated by reactive oxygen species (ROS) that accumulate as a result of oncogene activation [8], or by hyper-replication of DNA caused by sustained oncogenic signals [9,10]. OIS induction is also accompanied by accumulation of senescence-associated heterochromatic foci (SAHFs), which recruit Rb and heterochromatin proteins to silence stably the expression of E2F target genes that are necessary for cell proliferation [11]. These changes in chromatin brought about by SAHF formation are believed to mediate the irreversibility of OIS. Moreover, like replicative senescence, OIS is characterized by the SASP, referring to increased expression and secretion of inflammatory cytokines, chemokines, growth factors, proteases, and other proteins in senescent cells [12]. The SASP factors are crucial for the initiation and maintenance of senescence in a cell autonomous fashion [13–16], and some of them signal the immune system to clear senescent cells in vivo [17,18]. Some of the SASP factors are upregulated at the mRNA level by the transcription factors nuclear Trends in Biochemical Sciences xx (2014) 1–9

1

TIBS-1055; No. of Pages 9

Review factor k-light-chain-enhancer of activated B cells (NF-kB) and CCAAT-enhancer-binding protein b (C/EBPb) [13,15,19]. In this review, we discuss the molecular mechanisms and signal transduction pathways for OIS that have emerged from recent studies, focusing on the roles of the p38 MAPK and the PI3K/AKT/mTOR pathways. OIS and p38 MAPK pathway The p38 MAPK pathway was initially identified as a mediator of inflammation and stress responses (Box 1). Recent studies have indicated that the p38 pathway also mediates OIS and tumor suppression (Figure 1).

Trends in Biochemical Sciences xxx xxxx, Vol. xxx, No. x

Several studies have indicated that oncogenic ras-induced senescence is also mediated by Raf through the MAPK/ERK kinase (MEK)–ERK MAPK pathway [7,26,27]. Constitutively active mutants of Raf-1 and MEK1 induce senescence; consistently, inhibition of this pathway by chemical inhibitors of MEK1, dominant negative constructs of MEK1, or shRNA for ERK2 disrupts oncogenic ras-induced senescence. Further investigation has revealed that the MKK3/6-p38 pathway acts downstream of the Raf–MEK–ERK cascade to mediate rasinduced OIS [20,21] (Figure 1). Constitutively active MEK induced MKK3/6-p38 activation and senescence,

Sequential activation of the extracellular signalregulated kinase (ERK) and p38 MAPK pathways in OIS Multiple studies have revealed a major role of the p38 pathway in OIS induced by oncogenic ras or its downstream effector raf-1 [20–23]. The protein kinase activity of p38 is stimulated by oncogenic Ha-rasV12 or active raf-1 through its upstream kinases MAPK kinase (MKK)3 and MKK6 in human and murine fibroblasts, coinciding with induction of senescence [20–23]. Constitutive activation of p38 by ectopically expressed active mutants of MKK3 or MKK6 causes senescence, whereas inhibition of p38 activity by the pharmacological p38 inhibitor SB203580, a stably expressed dominant-negative mutant of MKK3 or MKK6, or short hairpin (sh)RNA-mediated knockdown of p38 expression overrides senescence induction [20,21,24,25]. These studies strongly indicate a central role for p38 in OIS.

Oncogenic Ras

Raf

MEK1/2

ERK

ROS, DNA damage

MKK3/6 p38α

Box 1. p38 MAPK pathway The p38 pathway is one of the major MAPK pathways, and was initially identified as a mediator of inflammation and stress responses [65]. The essential role of this pathway in cellular responses to components of microorganisms, inflammatory cytokines, and environmental stresses has been well established. Four isoforms of p38 (p38a, p38b, p38g and p38d; also known as stress activated protein kinase (SAPK)2a, 2b, 3 and 4, respectively) exist in the mammalian genome; each encoded by a different gene. These isoforms differ in their tissue distributions, regulation by upstream stimuli, and selectivity for upstream regulatory kinase and phosphatases as well as downstream targets. Like the other MAPK pathways, the p38 signaling cascade involves sequential activation of MAP kinase kinase kinases (MAP3Ks), such as MAP three kinase (MTK), mixed-lineage kinase (MLK)2, MLK3, dual leucine zipper-bearing kinase (DLK), apoptosis signal-regulating kinase (ASK), and TGFb-activated protein kinase (TAK)1, and MKKs including MKK3, MKK6, and MKK4, which directly activate p38 through phosphorylation in a cell type- and stimulus-dependent manner. An MKK-independent mechanism for p38 activation has also been observed, in which interaction between p38 and a chaperone protein called TAK1-binding protein (TAB)1 leads to autophosphorylation and activation of p38 [66]. After induction, p38 activity can be dampened through downregulation by dual-specific MAP kinase phosphatases (MKP)-1, -4 and -5, or other phosphatases such as protein phosphatase 2C (PP2C) [65]. The functions of p38 are mediated by downstream substrates, including transcription factors and cell-cycle regulators, and a family of Ser/Thr protein kinases consisting of MAPK-activated protein kinase (MK)2 and 3, p38-regulated/activated kinase (PRAK/MK5), MAPK-interacting protein kinase (MNK)1, mitogen- and stress-activated protein kinase MSK)1 and MSK2, and casein kinase (CK)2 [67]. In addition to inflammation and stress responses, the p38 pathway is involved in the control of cell cycle and proliferation [68], and has been shown to mediate both tumor suppression and tumor promotion in a contextdependent manner [69,70]. 2

AP-1/Ets

S401 P

T182 P

HBP1

PRAK

p38γ

CHK1/CHK2 S33 P

p16INK4A

p38δ

P S37

p53

DNA damage response

p21WAF1

OIS

Tumorigenesis Ti BS

Figure 1. Oncogenic ras-induced senescence is mediated by sequential activation of the Raf–MEK–ERK and MKK3/6–p38 pathways. Oncogenic ras initially activates the Raf–MEK–ERK pathway. Persistent activation of the Raf–MEK–ERK pathway leads to accumulation of ROS and DNA damage, which activate the stress-induced MKK3/6–p38 pathway. Unique among the p38 isoforms, expression of p38d is stimulated by AP-1 and Ets transcription factors, which are activated by the Raf– MEK–ERK pathway in response to oncogenic ras. Three p38 isoforms, p38a, p38g, and p38d, mediate OIS through differential mechanisms. p38a induces transcription of p16INK4A by directly phosphorylating and activating HBP1 transcription factor, and activates a downstream substrate kinase PRAK, which in turn activates p53 by phosphorylating p53 at Ser37. p38g stimulates the activity of p53 by directly phosphorylating p53 at Ser33. Once activated, p53 induces the expression of its transcriptional target p21WAF1, which, together with p16INK4A, triggers senescence. p38d mediates OIS through a p53- and p16INK4A-indepdent mechanism, possibly by regulating the activity of two DNA-damage checkpoint kinases CHK1 and CHK2 and participating in ras-induced DNA damage responses. ERK, extracellular signal-regulated kinase; HBP, high mobility group boxcontaining protein; MAPK, mitogen-activated protein kinase; MEK, MAPK/ERK kinase; MKK, MAPK kinase; OIS, oncogene-induced senescence; PRAK, p38 regulated/activated protein kinase; ROS, reactive oxygen species.

TIBS-1055; No. of Pages 9

Review and induction of p38 activity by ras requires not only active MKK3/6, but also active MEK. More importantly, although the p38 inhibitor SB203580 disrupts OIS induced by oncogenic ras, active MEK1 and active MKK3/6, the MEK inhibitor U0126 only prevents senescence induced by ras and active MEK1, but not by active MKK3/6. These studies place MKK3/6 and p38 downstream of MEK. The mechanism by which MKK3/6–p38 is activated following MEK– ERK activation remains to be determined. A likely intermediate between MEK–ERK and MKK3/6–p38 is ROS, which are induced by ras and mediate both p38 activation and OIS [28–30]. Oncogenic ras-induced accumulation of intracellular ROS is ERK-dependent during senescence induction [31]. Furthermore, shRNA-mediated depletion of ROS-generating oxidases nicotinamide adenine dinucleotide phosphateoxidase (Nox)1 and Nox4 leads to inhibition of oncogenic ras-induced p38 activation and disruption of OIS [32]. These findings suggest that ROS may act downstream of ERK to mediate p38 activation during ras-induced senescence. Another possible candidate is DNA damage. Hyperproliferative signals generated by activated MEK–ERK may cause hyper-replication of DNA, generating DNA damage that is known to activate p38 and induce senescence [9,10,33]. Of note, the involvement of ROS and DNA damage in the sequential activation of the ERK and p38 pathways may not be mutually exclusive, because ROS is a known DNA-damaging agent. Specifically, it has been shown that oncogenic ras-induced ROS cause activation of DNA damage responses [34]. Another study has demonstrated a positive feedback loop between ROS production and DNA damage responses: DNA damage responses trigger mitochondrial dysfunction leading to enhanced ROS production through a signaling pathway involving p53, p21WAF1, growth arrest and DNA-damageinducible protein (Gadd)45a, p38 and transforming growth factor (TGF)b, and ROS contribute to the long-term maintenance of DNA-damage responses, which are essential for induction of senescence [35]. Differential roles of p38 isoforms in OIS The mammalian genome encodes four isoforms of p38: p38a, p38b, p38g and p38d (Box 1). Recent studies have demonstrated that these p38 isoforms play different roles in OIS [24,25]. Senescence induction by ras is mediated by p38a, p38g and p38d, but not p38b, although all four isoforms are activated in senescent cells. shRNA-mediated silencing of p38a, p38g, or p38d disrupts ras-induced senescence, whereas shRNAs for p38b have no effect. In addition, the constitutively active mutant of p38a, p38g, or p38d, but not that of p38b, induces senescence in primary human fibroblasts. p38a, p38g, and p38d seem to mediate OIS through different mechanisms (Figure 1). It was shown in an early study that Ser33 of p53 is a substrate of p38a in vitro, and that phosphorylation of Ser33 contributes to p53 activation upon DNA damage [36]. Consistent with this report, recombinant p38a, p38b, p38g, and p38d all can phosphorylate p53–Ser33 in vitro with similar efficiency [24,25]. However, in cells oncogenic ras-induced phosphorylation of p53–Ser33 appears to be mediated by p38g, but not the other isoforms [24,25]. When immunoprecipitated from

Trends in Biochemical Sciences xxx xxxx, Vol. xxx, No. x

senescent cells, only p38g, but not p38a or p38d, phosphorylates p53–Ser33. In addition, oncogenic ras-induced phosphorylation of p53–Ser33 and the subsequent increase in transcriptional activity of p53 (as measured by p21WAF1 expression and activity of a p53-responsive luciferase reporter) in senescent cells is greatly reduced by p38g shRNA, but not by shRNA for p38a or p38d. Moreover, the constitutively active form of p38g, but not that of p38a, p38b, or p38d, induces p53–Ser33 phosphorylation and p21WAF1 expression in cells. These findings indicate that during oncogenic ras-induced senescence in cells, phosphorylation of p53–Ser33 and activation of p53 is mainly mediated by p38g, but not the other isoforms. The mechanism underlying this discrepancy between the in vitro and in vivo activities of the p38 isoforms toward p53 is currently unknown. It is possible that during senescence induction in cells, the kinase activity of p38g toward p53 is enhanced as a result of post-translational modification or binding to a positive regulator. Alternatively, the p53 kinase activity of the other p38 isoforms may be repressed by post-translational modification or an associated inhibitory protein in cells. Although p38a does not contribute to p53 activation in ras-induced senescence, it is essential for oncogenic rasinduced expression of p16INK4A; another key senescence effector. p38a shRNA, but not p38g shRNA, abolishes induction of p16INK4A by ras [24]. The regulation of p16INK4A by p38a is likely to be mediated by the transcription factor high mobility group (HMG) box-containing protein (HBP)1. Without distinguishing the effects of different p38 isoforms, one study has shown that HBP1 is a direct substrate of p38, and phosphorylation of HBP1 at Ser401 by p38 leads to stabilization of the HBP1 protein [37]. The HBP1 protein level indeed increases in cells undergoing ras- or active MKK6 (MKK6E)-induced senescence, where p38 is activated [38,39]. HBP1 mediates senescence induction by the ras–p38 pathway through upregulation of p16INK4A, because HBP1 shRNAs abrogate senescence and p16INK4A induction by oncogenic ras or activated p38, whereas HBP1 overexpression induces senescence and p16INK4A expression [38,39]. Furthermore, HBP1 directly binds to the p16INK4A promoter and upregulates p16INK4A transcription during oncogenic ras-induced senescence, and oncogenic ras and MKK6E enhance the promoter activity of p16INK4A in a HBP1-dependent fashion [39]. These results indicate that p38 induces the transcription of p16INK4A by phosphorylating and activating the p16INK4A transcription factor HBP1 in ras-induced senescence. It will be interesting to investigate the possibility that HBP1 is specifically phosphorylated and activated by p38a to mediate p16INK4A induction. In contrast to p38a and p38g, p38d mediates oncogenic ras-induced senescence in a p53- and p16INK4A-independent manner [25]. Although p38d shRNA disrupts OIS and the constitutively active mutant of p38d triggers senescence, neither has any effect on p16INK4A expression, p53– Ser33 phosphorylation or p53 transcriptional activity. Interestingly, p38d shRNA reduces ras-induced activating phosphorylation of checkpoint kinase (CHK)1 and CHK2, two DNA-damage checkpoint kinases, suggesting that p38d may participate in the DNA-damage responses to 3

TIBS-1055; No. of Pages 9

Review oncogenic ras. However, how p38d regulates DNA damage responses in a p53- and p16INK4A-indendent fashion remains unclear. p38d is unique among the p38 isoforms in that its expression is upregulated during ras-induced senescence [25]. Oncogenic ras induces the Raf-1–MEK– ERK pathway, which, in turn, activates the activator protein (AP)-1 and v-Ets avian erythroblastosis virus E26 oncogene homolog (Ets) transcription factors that are bound to the p38d promoter, leading to increased transcription of p38d. Therefore, induction of the prosenescent function of p38d by oncogenic ras is achieved through two mechanisms: transcriptional activation by Raf-1–MEK–ERK–AP-1–Ets pathway, which the increases the cellular concentration of the p38d protein, and post-translational modification by MKK3/6, which stimulates the enzymatic activity of p38d. Role of a p38 downstream substrate kinase PRAK in OIS Besides regulating the activity of p53 through direct phosphorylation, p38 also mediates ras-induced senescence through a downstream Ser/Thr protein kinase called p38 regulated/activated protein kinase (PRAK), or MAPK-activated protein kinase 5 (MAPKAPK5 or MK5) [40] (Figure 1). PRAK, which is activated by ras in a p38-dependent manner, is essential for senescence induction by ras in primary human and murine fibroblasts. In mice, senescence induction is also compromised by PRAK deficiency in skin papillomas induced by 7,12-dimethylbenz(a)anthracene (DMBA); an environmental carcinogen that prompts oncogenic ras mutations. The function of PRAK in OIS is at least in part mediated by p53. PRAK is essential for ras-induced transcriptional activity of p53. Upon activation by the Ras–p38 cascade, PRAK directly phosphorylates p53 at Ser37; a residue required for p53 to mediate ras-induced senescence. This report also demonstrates that in addition to the PRAK phosphorylation site Ser37, other p53 residues that are phosphorylated during oncogenic ras-induced senescence, including the p38 phosphorylation site Ser33, are important for the ability of p53 to mediate OIS [40]. Therefore, activation of p53 during OIS requires the coordinated phosphorylation of different residues by multiple components of the p38 pathway. The identity of the p38 isoform responsible for PRAK phosphorylation in OIS is unclear; although in vitro PRAK is mainly activated by p38a and p38b, but not by p38g and p38d [41]. This raises a possibility that PRAK is activated primarily by p38a in OIS, as p38b is dispensable for OIS. Post-translational modification cascade involving p38, Tip60 and PRAK in OIS In an attempt to delineate the pathway mediating the function of PRAK in OIS, a yeast-2-hybrid screen was performed to search for PRAK-interacting proteins [42]. This screen led to the identification of Tat interactive protein, 60 kDa (Tip60), a member of the monocytic leukemia zinc finger (MOZ), YBF2, SAS2 and Tip60 (MYST) family of acetyltransferases. Tip60 has been previously implicated in multiple cellular processes, including DNA-damage responses, apoptosis, and cell proliferation [43,44], and has been shown to function as a tumor suppressor [45]. This study has demonstrated that Tip60 is essential for 4

Trends in Biochemical Sciences xxx xxxx, Vol. xxx, No. x

Oncogenic Ras

MKK3/6 p38α

p38δ

T158 P Tip60

T182 P PRAK Ac K364 P S37

p53

OIS Ti BS

Figure 2. Post-translational modification cascade that regulates OIS. Upon activation by oncogenic ras, p38a and p38d induce acetyltransferase activity of Tip60 through phosphorylation of Thr158. Activated Tip60, which directly interacts with PRAK, induces protein kinase activity of PRAK through acetylation of Lys364. Phosphorylation of PRAK at Thr182 by p38a is required for the optimal acetylation of PRAK by Tip60. Activated PRAK in turn phosphorylates p53 at Ser37, thereby stimulating the activity of p53 in OIS induction. MKK, mitogen-activated protein kinase kinase; OIS, oncogene-induced senescence; PRAK, p38 regulated/activated protein kinase; Tip60, Tat interactive protein, 60 kDa.

ras-induced senescence, thus providing a mechanism underlying the tumor-suppressing activity of Tip60 [42]. Further investigation has revealed a post-translational modification cascade involving p38, Tip60, and PRAK; three proteins essential for OIS (Figure 2). Upon activation by ras, p38 (likely p38a and p38d) induces the acetyltransferase activity of Tip60 through phosphorylation of Thr158; activated Tip60, which directly interacts with PRAK, in turn induces the protein kinase activity of PRAK through acetylation of Lys364 in a manner that depends on phosphorylation of both Tip60 and PRAK by p38. These post-translational modifications are crucial for the prosenescent function of Tip60 and PRAK, because substitutions that disrupt these modifications (T158A in Tip60 and K364R in PRAK) render Tip60 and PRAK incapable of restoring ras-induced senescence in cells expressing shRNAs for Tip60 and PRAK, respectively. These findings have thus defined a novel signaling circuit within the p38 pathway, which mediates OIS. The impact of this circuit on OIS induction is at least partly achieved through regulation of p53 activity, because PRAK-mediated p53–Ser37 phosphorylation in response to oncogenic ras is greatly reduced in cells with Tip60 knockdown. Of note, it has been shown previously that Tip60 regulates the activity of p53 by directly acetylating p53 at Lys120 [46,47]. Oncogenic ras indeed induces acetylation of p53–Lys120; however, rasinduced p53–Lys120 acetylation is not reduced by Tip60 shRNA [42], suggesting that p53–Lys120 acetylation in OIS may be mediated by a different acetyltransferase. Tumor-suppressing function of the p38 pathway The role of p38 MAPK in OIS suggests that this pathway may be involved in the suppression of tumor development.

TIBS-1055; No. of Pages 9

Review Indeed, the tumor-suppressing function of several p38 pathway components has been demonstrated in mouse cancer models in vivo. Conditional deletion of p38a in adult mice enhances both initiation and progression of K-RasG12V-induced lung cancer due to hyperproliferation of lung epithelium and reduced differentiation of lung progenitor cells [48]. An independent study has demonstrated that, compared to wild-type animals, mice with liver-specific deletion of p38a show accelerated diethylnitrosamine (DEN)-phenobarbital (Pb)-induced liver tumor development and increased DEN-induced hepatocyte proliferation, which correlates with upregulation of c-Jun N-terminal kinase (JNK)–cJun activity [49]. Notably, simultaneous deletion of cJun in liver abolishes the enhancement of liver tumor induction and tumor cell proliferation observed in p38adeficient mice, indicating that p38a inhibits the JNK–cJun pathway to control liver cell proliferation and suppress cancer development. Taken together, these results have demonstrated a tumor-suppressing function of p38a in vivo. However, it is unclear whether p38a deficiency promotes tumorigenesis by disrupting OIS in these studies. Another study has shown that the p38 downstream kinase PRAK also suppresses tumorigenesis in vivo. PRAK deletion renders mice prone to skin papilloma induction by DMBA, which induces activating ras mutations, and accelerates lymphomagenesis in Em-N-RasG12D transgenic mice, which specifically express an activated N-ras transgene in hematopoietic cells [40,50]. In these models, enhanced tumorigenesis is observed in both PRAK / and PRAK+/ mice. Thus, the tumor-suppressing function of PRAK is haploinsufficient. Acceleration of DMBA-induced skin carcinogenesis in PRAK-deficient mice is accompanied by compromised senescence induction, suggesting that the tumor-suppressing effect of PRAK is achieved via its ability to mediate OIS. Modulation of p38 activity by manipulating its upstream regulators can also affect tumor development in vivo. Deletion of the p38 phosphatase wild type p53induced phosphatase 1 (Wip1), which results in p38 activation, inhibits mammary tumorigenesis in mice with mammary-gland-specific expression of oncogene erbB2 or v-Ha-ras; both driven by the mouse mammary tumor virus (MMTV) promoter [51]. Inactivation of p38 by SB203580 treatment restores MMTV-erbB2-induced mammary tumor formation in Wip1 / mice. In a reciprocal experiment, mice with targeted expression of Wip1 in the breast epithelium are prone to mammary tumor induction by the MMTV-erbB2 transgene, and the enhancement of tumorigenesis by Wip1 is eliminated when a constitutively active MKK6 is introduced into these mice [52]. These results indicate that p38 is the target for the tumor-promoting function of Wip1 during breast tumorigenesis in MMTVerbB2 mice. In addition, deletion of Gadd45a, a p38 binding partner and activator, accelerates MMTV-ras-induced mammary tumorigenesis [53], partly due to decreased OIS as a result of impaired p38 activation. Consistent with the tumor-suppressing function of p38, this pathway is downregulated in human cancers. In a study surveying 20 hepatocellular carcinoma patients, the p38 and MKK6 kinase activities are significantly lower in tumorous lesions as compared to the paired nontumorous

Trends in Biochemical Sciences xxx xxxx, Vol. xxx, No. x

tissues, and larger tumors exhibit lower levels of p38 and MKK6 activity than the smaller tumors [54]. p38a expression is also lower in human lung tumors as compared with normal lung tissues [48]. The p38 phosphatase Wip1 is frequently activated through amplification in human breast cancer, resulting in impaired p38 activation in these cells [22,55]. Furthermore, glutathione S-transferase Mu 1 (Gstm1), a member of the glutathione S-transferase family that inhibit p38 activation by oxidative stress, is overexpressed in multiple types of cancers [31]. In summary, recent studies have demonstrated a novel function of the p38 pathway in tumor suppression, which is at least partly attributed to the ability of this pathway to mediate OIS. OIS and PI3K/AKT/mTOR pathway The PI3K/AKT/mTOR pathway is frequently activated in human cancer (Box 2). Alterations that lead to constitutive activation of this pathway in cancer include genetic and epigenetic inactivation of phosphatase and tensin homolog (PTEN), overexpression or activating mutation of PI3K (PIK3CA) and overexpression of AKT [56]. Recent studies have indicated that activation of this pathway through multiple components lead to induction of OIS, and have begun to reveal the downstream effectors (Figure 3A). It was reported initially that a constitutively active, myristoylated form of AKT induces OIS in primary murine fibroblasts and multiple lines of primary cultured human endothelial cells via a p53/p21WAF1-dependent pathway [57]. This is mediated by downregulation of the forkhead transcription factor FOXO3a, which is an AKT downstream substrate. FOXO3a upregulates the transcription of radical scavenger genes, such as manganese superoxide dismutase (MnSOD), which protect cells from oxidative Box 2. PI3K/AKT/mTOR signaling pathway The PI3K/AKT/mTOR pathway is one of the most important intracellular pathways, and is frequently activated in human cancers. Under normal physiological conditions, the activity of the PI3K/AKT/mTOR pathway is tightly controlled [56,71,72]. Upon activation by receptor tyrosine kinases or G-protein-coupled receptors, PI3K is translocated to the plasma membrane, leading to phosphorylation of phosphatidylinositol 4,5-bisphosphate (PIP2) to phosphatidylinositol 3,4,5-triphosphate (PIP3). This step is negatively regulated by the PTEN phosphatase, which dephosphorylates PIP3 to PIP2. PIP3 recruits the serine–threonine kinase AKT and its activating kinase 3-phosphoinositide-dependent kinase (PDK)1 to the plasma membrane, where PDK1 activates AKT by phosphorylating threonine 308. Activated AKT phosphorylates tuberous sclerosis complex (TSC)2 and thereby inhibits the GTPase-activating protein (GAP) activity of the TSC1/TSC2 complex toward Ras homolog enriched in brain (Rheb). This allows the GTP-bound, active form of Rheb to accumulate and activate mTOR. mTOR forms either mTOR complex 1 (mTORC1) with the Raptor protein, or mTORC2 with the Rictor protein. Activation of mTORC1 results in increased protein translation through inhibitory phosphorylation of eukaryotic translational initiation factor eIF4E-binding protein 1 (4EBP1), which in turn inhibits eIF4E, and activating phosphorylation of p70S6 kinase (S6K), which in turn phosphorylates ribosomal S6 protein. Activated mTORC2 serves as a positive feedback loop leading to additional activating phosphorylation of AKT. AKT also regulates transcription by inducing the phosphorylation-dependent degradation of forkhead box O (FOXO) transcription factor and inhibitory phosphorylation of glycogen synthase kinase (GSK)3b, which negatively regulates the function of c-Jun and Myc transcription factors. 5

TIBS-1055; No. of Pages 9

Review

Trends in Biochemical Sciences xxx xxxx, Vol. xxx, No. x

(A)

Complete loss of PTEN

TSC1

Oncogenic Ras

(B)

PI3K

Paral loss of PTEN

AKT

PI3K

TSC2

FOXO3a

p19ARF

RHEB

MnSOD

MDM2

mTOR

ROS

AKT

OIS

p53

p21WAF1

OIS Ti BS

Figure 3. Role of PI3K/AKT/mTOR pathway in OIS. (A) Complete loss of PTEN or ectopic expression of oncogenic ras or constitutively active forms of PI3K or AKT leads to strong activation of the PI3K/AKT/mTOR pathway (purple, thick arrows). Activated mTOR stimulates the translation of p53, which in turn stimulates p21WAF1 expression and triggers OIS. Strong activation of AKT also inhibits the FOXO3a transcription factor that upregulates the transcription of a radical scavenger gene MnSOD, leading to decreased MnSOD expression, accumulation of ROS, and activation of the p51/p21WAF1 cascade that mediates OIS. In addition, complete loss of PTEN induces the expression of p19ARF, an inhibitor of MDM2 that serves as a negative regulator of p53, resulting in increased expression and activity of p53. (B) Partial loss of PTEN leads to moderate activation of the PI3K/AKT pathways (thin arrows), which disrupts OIS. FOXO3a, forkhead box O3a; MDM2, murine double minute 2; MnSOD, manganese superoxide dismutase; mTOR, mammalian target of rapamycin; OIS, oncogene-induced senescence; PI3K, phosphoinositide 3-kinase; PTEN, phosphatase and tensin homolog; RHEB, Ras homolog enriched in brain; ROS, reactive oxygen species; TSC, tuberous sclerosis complex.

damage. Active AKT inhibits the transcriptional activity of FOXO3a and thereby downregulates MnSOD, leading to an increased level of ROS that induces activation of the p53/p21WAF1 pathway and senescence. In addition to AKT activation, loss of PTEN also triggers p53-dependent OIS in primary murine fibroblasts [58]. Complete loss of PTEN induces the expression of p19ARF, an inhibitor of the p53-specific E3 ubiquitin-protein ligase murine double minute 2 (MDM2). This leads to stabilization of the p53 protein and increased expression of p21WAF1, resulting in senescence. More importantly, homozygous deletion of PTEN in prostatic epithelium in mice induced premalignant lesions containing large numbers of senescent cells, which is accompanied by AKT activation and upregulation of p19ARF, p53, and p21WAF1 proteins. Loss of p53 disrupts senescence in the prostate and enhances the development of invasive prostate cancer in PTEN-null mice. These results indicate that loss of PTEN, through the resulting activation of the PI3K/AKT pathway, triggers a p19ARF/p53/p21WAF1-dependent senescence program that suppresses cancer development in vivo. Moreover, a cancer-derived, constitutively active mutant of the catalytic subunit of PI3K (PIK3CA)–E545K, also induces senescence in primary human fibroblasts [59]. These 6

findings indicate that activation of the PI3K/AKT/mTOR pathway through multiple components can trigger OIS (Figure 3A). Further supporting the importance of the PI3K/AKT/ mTOR pathway in OIS, mTOR is required for oncogenic ras-induced senescence in primary human fibroblasts [60]. Inhibition of the mTORC1 complex by the mTORC1-specific inhibitor rapamycin or shRNAs against mTOR or regulatory-associated protein of mTOR (Raptor) delays both replicative and oncogenic ras-induced senescence, whereas inhibition of the mTORC2 complex by rapamycin-insensitive companion of mTOR (Rictor) shRNA has no effect. In addition, rapamycin treatment disrupts senescence and p53 accumulation induced by loss of PTEN or constitutively active AKT in primary human and murine cells [59,61]. Treatment with a rapamycin analogue RAD001 also reduces induction of senescence and p53 expression in PTEN-null prostate epithelium in mice [61]. Moreover, PTEN loss induces the translation of the p53 gene in primary murine fibroblasts, presumably as a result of the upregulation of overall gene translation by activated mTOR [61]. These findings indicate that mTORC1, but not mTORC2, is an essential component in the OIS pathway.

TIBS-1055; No. of Pages 9

Review Unlike classical OIS, such as that induced by HarasV12, Mos and CDK6, senescence induced by PTEN loss or activated AKT is not accompanied by DNA-damage responses [59,61]. Defects in the DNA-damage response pathway abrogate Ha-rasV12-induced senescence, but have no effect on senescence induced by PTEN loss [61]. In addition, whereas Ha-rasV12-induced senescence is preceded by an initial period of enhanced cell proliferation [3,7], PTEN loss- and AKT-induced senescence occurs rapidly without an early phase of hyperproliferation and does not rely on DNA replication [59,61]. These results suggest that different oncogenes induce senescence through overlapping, but distinct pathways. Interestingly, a recent study has indicated that the nuclear function of PTEN is required for homologous recombination repair of DNA double-strand breaks induced by DNA-damaging agents, and that cells lacking PTEN display persistent DNA damage responses due to defects in DNA repair [62]. It is possible that the DNA repair function of PTEN only becomes essential in the face of genotoxic stress, and thus loss of PTEN in the absence of DNA-damaging agents is not sufficient to trigger DNA damage responses. The role of the PI3K/AKT/mTOR pathway in OIS is challenged by a recent report that instead of mediating senescence, activation of this pathway abrogates senescence induction by BRAFV600E [63]. shRNA-mediated depletion of PTEN, which induces AKT activation, disrupts BRAFV600E-induced senescence in both primary human fibroblasts and primary human melanocytes. Using a BRAFV600E knock-in mouse model with melanocyte-specific expression of BRAFV600E that develops nevus-like lesions characterized by melanocytic hyperplasia, the authors have found that injection of a lentivirus carrying PTEN shRNA into the nevi leads to tumor formation, suggesting that PTEN depletion allows BRAFV600Eexpressing nevus cells to resume proliferation. However, whether the tumor formation is indeed due to disruption of senescence in nevi was not shown. Although one cannot rule out the contribution of tissuespecific effects and differences in the specific oncogenes used to induce senescence, one major difference between these studies demonstrating opposite roles of PTEN loss in senescence is the degree of PTEN inactivation. In prostate cancer studies [58,61], senescence is induced by homozygous, but not heterozygous, deletion of PTEN both in primary fibroblasts in vitro and in prostate epithelium in vivo. Treatment with a PTEN inhibitor VO-OHpic [V(=O)(H2O)(OHpic)2] induces senescence in PTEN+/ cells, whereas the wild-type cells are unaffected [61], suggesting that PTEN activity has to be significantly reduced by >50% in order for senescence to occur. By contrast, in the study demonstrating the anti-senescence function of PTEN loss [63], PTEN was knocked down by shRNAs, which only partially inhibited PTEN expression. Indeed, the authors failed to overexpress activated PI3K or AKT family members in melanocytes, raising a possibility that strong PI3K/AKT signaling might induce senescence. Thus, it is likely that the outcome of PI3K/AKT/mTOR pathway activation with regard to OIS depends on the dosage of the PTEN gene, and thus the signaling strength of the PI3K/AKT/mTOR pathway (Figure 3). Although

Trends in Biochemical Sciences xxx xxxx, Vol. xxx, No. x

strong activation of this pathway is prosenescent and tumor suppressing (Figure 3A), moderate activation abrogates senescence induction by other oncogenes leading to enhanced tumorigenesis (Figure 3B). Concluding remarks Mounting evidence has indicated that OIS is an important tumor-suppressing defense response that restricts the progression of benign lesions to malignancy in vivo. The molecular mechanisms underlying OIS have begun to be unveiled. There is solid evidence that OIS is almost invariably enforced by the p53/p21WAF1 and/or p16INK4A tumorsuppressor pathways. However, the signaling pathways mediating the activation of p53 and p16INK4A can vary depending on the cell type and the OIS inducer. It should also be noted that although the p53/p21WAF1 and p16INK4A pathways are major regulators of OIS, most of the data were obtained using fibroblasts, and the situation might be more complex in other cell types [64]. In addition, activation of p53 and p16INK4A alone is not sufficient to confer the irreversibility of OIS. Additional mechanisms, such as DNA-damage responses, chromatin remodeling through SAHF formation, and the SASP are required for the initiation and maintenance of the irreversible growth arrest and other phenotypes of OIS. Recent studies have demonstrated a role of the p38 MAPK and PI3K/AKT/mTOR pathways in these processes leading to OIS. These findings indicate that OIS is not mediated by a simple, linear pathway, but by an intricate signaling network that is often context dependent. Further investigations are needed to identify novel players in OIS, and to define the crosstalk and cooperation among these different signaling pathways involved in OIS. A better understanding of the mechanisms that mediate OIS may lead to identification of new targets for cancer therapies based on restoration of OIS in cancer cells. Acknowledgments We thank the Sun laboratory for stimulating discussions. The Sun laboratory is supported by the National Institutes of Health (CA106768, CA131231 and CA172115).

References 1 Hayflick, L. (2003) Living forever and dying in the attempt. Exp. Gerontol. 38, 1231–1241 2 Shay, J.W. and Wright, W.E. (2005) Senescence and immortalization: role of telomeres and telomerase. Carcinogenesis 26, 867–874 3 Serrano, M. et al. (1997) Oncogenic ras provokes premature cell senescence associated with accumulation of p53 and p16INK4a. Cell 88, 593–602 4 Wei, S. et al. (1999) Expression of catalytically active telomerase does not prevent premature senescence caused by overexpression of oncogenic Ha-Ras in normal human fibroblasts. Cancer Res. 59, 1539–1543 5 Suram, A. et al. (2012) Oncogene-induced telomere dysfunction enforces cellular senescence in human cancer precursor lesions. EMBO J. 31, 2839–2851 6 Courtois-Cox, S. et al. (2008) Many roads lead to oncogene-induced senescence. Oncogene 27, 2801–2809 7 Lin, A.W. et al. (1998) Premature senescence involving p53 and p16 is activated in response to constitutive MEK/MAPK mitogenic signaling. Genes Dev. 12, 3008–3019 8 Lee, A.C. et al. (1999) Ras proteins induce senescence by altering the intracellular levels of reactive oxygen species. J. Biol. Chem. 274, 7936– 7940 7

TIBS-1055; No. of Pages 9

Review 9 Bartkova, J. et al. (2006) Oncogene-induced senescence is part of the tumorigenesis barrier imposed by DNA damage checkpoints. Nature 444, 633–637 10 Di Micco, R. et al. (2006) Oncogene-induced senescence is a DNA damage response triggered by DNA hyper-replication. Nature 444, 638–642 11 Adams, P.D. (2007) Remodeling of chromatin structure in senescent cells and its potential impact on tumor suppression and aging. Gene 397, 84–93 12 Freund, A. et al. (2010) Inflammatory networks during cellular senescence: causes and consequences. Trends Mol. Med. 16, 238–246 13 Kuilman, T. et al. (2008) Oncogene-induced senescence relayed by an interleukin-dependent inflammatory network. Cell 133, 1019– 1031 14 Wajapeyee, N. et al. (2008) Oncogenic BRAF induces senescence and apoptosis through pathways mediated by the secreted protein IGFBP7. Cell 132, 363–374 15 Acosta, J.C. et al. (2008) Chemokine signaling via the CXCR2 receptor reinforces senescence. Cell 133, 1006–1018 16 Yang, G. et al. (2006) The chemokine growth-regulated oncogene 1 (Gro-1) links RAS signaling to the senescence of stromal fibroblasts and ovarian tumorigenesis. Proc. Natl. Acad. Sci. U.S.A. 103, 16472– 16477 17 Xue, W. et al. (2007) Senescence and tumour clearance is triggered by p53 restoration in murine liver carcinomas. Nature 445, 656–660 18 Krizhanovsky, V. et al. (2008) Senescence of activated stellate cells limits liver fibrosis. Cell 134, 657–667 19 Freund, A. et al. (2011) p38MAPK is a novel DNA damage responseindependent regulator of the senescence-associated secretory phenotype. EMBO J. 30, 1536–1548 20 Wang, W. et al. (2002) Sequential activation of the MEK-extracellular signal-regulated kinase and MKK3/6-p38 mitogen-activated protein kinase pathways mediates oncogenic ras-induced premature senescence. Mol. Cell. Biol. 22, 3389–3403 21 Iwasa, H. et al. (2003) Mitogen-activated protein kinase p38 defines the common senescence-signalling pathway. Genes Cells 8, 131–144 22 Bulavin, D.V. et al. (2002) Amplification of PPM1D in human tumors abrogates p53 tumor-suppressor activity. Nat. Genet. 31, 210–215 23 Bulavin, D.V. et al. (2003) Loss of oncogenic H-ras-induced cell cycle arrest and p38 mitogen-activated protein kinase activation by disruption of Gadd45a. Mol. Cell. Biol. 23, 3859–3871 24 Kwong, J. et al. (2009) p38alpha and p38gamma mediate oncogenic rasinduced senescence through differential mechanisms. J. Biol. Chem. 284, 11237–11246 25 Kwong, J. et al. (2013) Induction of p38delta expression plays an essential role in oncogenic ras-induced senescence. Mol. Cell. Biol. 33, 3780–3794 26 Zhu, J. et al. (1998) Senescence of human fibroblasts induced by oncogenic Raf. Genes Dev. 12, 2997–3007 27 Shin, J. et al. (2013) Depletion of ERK2 but not ERK1 abrogates oncogenic Ras-induced senescence. Cell. Signal. 25, 2540–2547 28 Zdanov, S. et al. (2006) Identification of p38MAPK-dependent genes with changed transcript abundance in H2O2-induced premature senescence of IMR-90 hTERT human fibroblasts. FEBS Lett. 580, 6455–6463 29 Colavitti, R. and Finkel, T. (2005) Reactive oxygen species as mediators of cellular senescence. IUBMB Life 57, 277–281 30 Nicke, B. et al. (2005) Involvement of MINK, a Ste20 family kinase, in Ras oncogene-induced growth arrest in human ovarian surface epithelial cells. Mol. Cell 20, 673–685 31 Dolado, I. et al. (2007) p38alpha MAP kinase as a sensor of reactive oxygen species in tumorigenesis. Cancer Cell 11, 191–205 32 Kodama, R. et al. (2013) ROS-generating oxidases Nox1 and Nox4 contribute to oncogenic Ras-induced premature senescence. Genes Cells 18, 32–41 33 Wang, X. et al. (2000) Involvement of the MKK6-p38gamma cascade in gamma-radiation-induced cell cycle arrest. Mol. Cell. Biol. 20, 4543– 4552 34 Ogrunc, M. et al. (2014) Oncogene-induced reactive oxygen species fuel hyperproliferation and DNA damage response activation. Cell Death Differ. http://dx.doi.org/10.1038/cdd.2014.16 35 Passos, J.F. et al. (2010) Feedback between p21 and reactive oxygen production is necessary for cell senescence. Mol. Syst. Biol. 6, 347 8

Trends in Biochemical Sciences xxx xxxx, Vol. xxx, No. x

36 Bulavin, D.V. et al. (1999) Phosphorylation of human p53 by p38 kinase coordinates N-terminal phosphorylation and apoptosis in response to UV radiation. EMBO J. 18, 6845–6854 37 Xiu, M. et al. (2003) The transcriptional repressor HBP1 is a target of the p38 mitogen-activated protein kinase pathway in cell cycle regulation. Mol. Cell. Biol. 23, 8890–8901 38 Zhang, X. et al. (2006) The HBP1 transcriptional repressor participates in RAS-induced premature senescence. Mol. Cell. Biol. 26, 8252–8266 39 Li, H. et al. (2010) Transcriptional factor HBP1 targets P16(INK4A), upregulating its expression and consequently is involved in Rasinduced premature senescence. Oncogene 29, 5083–5094 40 Sun, P. et al. (2007) PRAK is essential for ras-induced senescence and tumor suppression. Cell 128, 295–308 41 New, L. et al. (1998) PRAK, a novel protein kinase regulated by the p38 MAP kinase. EMBO J. 17, 3372–3384 42 Zheng, H. et al. (2013) A posttranslational modification cascade involving p38, Tip60, and PRAK mediates oncogene-induced senescence. Mol. Cell 50, 699–710 43 Sapountzi, V. et al. (2006) Cellular functions of TIP60. Int. J. Biochem. Cell Biol. 38, 1496–1509 44 Squatrito, M. et al. (2006) Tip60 in DNA damage response and growth control: many tricks in one HAT. Trends Cell Biol. 16, 433–442 45 Gorrini, C. et al. (2007) Tip60 is a haplo-insufficient tumour suppressor required for an oncogene-induced DNA damage response. Nature 448, 1063–1067 46 Charvet, C. et al. (2011) Phosphorylation of Tip60 by GSK-3 determines the induction of PUMA and apoptosis by p53. Mol. Cell 42, 584–596 47 Tang, Y. et al. (2006) Tip60-dependent acetylation of p53 modulates the decision between cell-cycle arrest and apoptosis. Mol. Cell 24, 827–839 48 Ventura, J.J. et al. (2007) p38alpha MAP kinase is essential in lung stem and progenitor cell proliferation and differentiation. Nat. Genet. 39, 750–758 49 Hui, L. et al. (2007) p38alpha suppresses normal and cancer cell proliferation by antagonizing the JNK-c-Jun pathway. Nat. Genet. 39, 741–749 50 Yoshizuka, N. et al. (2012) PRAK suppresses oncogenic ras-induced hematopoietic cancer development by antagonizing the JNK pathway. Mol. Cancer Res. 10, 810–820 51 Bulavin, D.V. et al. (2004) Inactivation of the Wip1 phosphatase inhibits mammary tumorigenesis through p38 MAPK-mediated activation of the p16(Ink4a)-p19(Arf) pathway. Nat. Genet. 36, 343–350 52 Demidov, O.N. et al. (2006) The role of the MKK6/p38 MAPK pathway in Wip1-dependent regulation of ErbB2-driven mammary gland tumorigenesis. Oncogene 26, 2502–2506 53 Tront, J.S. et al. (2006) Gadd45a suppresses Ras-driven mammary tumorigenesis by activation of c-Jun NH2-terminal kinase and p38 stress signaling resulting in apoptosis and senescence. Cancer Res. 66, 8448–8454 54 Iyoda, K. et al. (2003) Involvement of the p38 mitogen-activated protein kinase cascade in hepatocellular carcinoma. Cancer 97, 3017–3026 55 Rauta, J. et al. (2006) The serine-threonine protein phosphatase PPM1D is frequently activated through amplification in aggressive primary breast tumours. Breast Cancer Res. Treat. 95, 257–263 56 Engelman, J.A. et al. (2006) The evolution of phosphatidylinositol 3kinases as regulators of growth and metabolism. Nat. Rev. Genet. 7, 606–619 57 Miyauchi, H. et al. (2004) Akt negatively regulates the in vitro lifespan of human endothelial cells via a p53/p21-dependent pathway. EMBO J. 23, 212–220 58 Chen, Z. et al. (2005) Crucial role of p53-dependent cellular senescence in suppression of Pten-deficient tumorigenesis. Nature 436, 725–730 59 Astle, M.V. et al. (2012) AKT induces senescence in human cells via mTORC1 and p53 in the absence of DNA damage: implications for targeting mTOR during malignancy. Oncogene 31, 1949–1962 60 Kolesnichenko, M. et al. (2012) Attenuation of TORC1 signaling delays replicative and oncogenic RAS-induced senescence. Cell Cycle 11, 2391–2401 61 Alimonti, A. et al. (2010) A novel type of cellular senescence that can be enhanced in mouse models and human tumor xenografts to suppress prostate tumorigenesis. J. Clin. Invest. 120, 681–693 62 Bassi, C. et al. (2013) Nuclear PTEN controls DNA repair and sensitivity to genotoxic stress. Science 341, 395–399

TIBS-1055; No. of Pages 9

Review 63 Vredeveld, L.C. et al. (2012) Abrogation of BRAFV600E-induced senescence by PI3K pathway activation contributes to melanomagenesis. Genes Dev. 26, 1055–1069 64 Bianchi-Smiraglia, A. and Nikiforov, M.A. (2012) Controversial aspects of oncogene-induced senescence. Cell Cycle 11, 4147–4151 65 Ono, K. and Han, J. (2000) The p38 signal transduction pathway: activation and function. Cell. Signal. 12, 1–13 66 Lu, G. et al. (2006) TAB-1 modulates intracellular localization of p38 MAP kinase and downstream signaling. J. Biol. Chem. 281, 6087–6095 67 Shi, Y. and Gaestel, M. (2002) In the cellular garden of forking paths: how p38 MAPKs signal for downstream assistance. Biol. Chem. 383, 1519–1536

Trends in Biochemical Sciences xxx xxxx, Vol. xxx, No. x

68 Bulavin, D.V. and Fornace, A.J., Jr (2004) p38 MAP kinase’s emerging role as a tumor suppressor. Adv. Cancer Res. 92, 95–118 69 Han, J. and Sun, P. (2007) The pathways to tumor suppression via route p38. Trends Biochem. Sci. 32, 364–371 70 Yoshizuka, N. et al. (2012) A novel function of p38-regulated/activated kinase in endothelial cell migration and tumor angiogenesis. Mol. Cell. Biol. 32, 606–618 71 Zhao, L. and Vogt, P.K. (2008) Class I PI3K in oncogenic cellular transformation. Oncogene 27, 5486–5496 72 Polivka, J., Jr and Janku, F. (2013) Molecular targets for cancer therapy in the PI3K/AKT/mTOR pathway. Pharmacol. Ther. 142, 164–175

9

mTOR pathways in oncogene-induced senescence.

Oncogene-induced senescence (OIS) is a tumor-suppressing response that must be disrupted for cancer to develop. Mechanistic insights into OIS have beg...
664KB Sizes 2 Downloads 4 Views